Identification of an Orally Bioavailable, Potent, and Selective Inhibitor

Jun 25, 2010 - Angus M. MacLeod,. †. Andrew Pike,. †. Suzanne Wood,. †. Steve Thomas, ... H. Donald Burns,. ‡. Waisi Eng,. ‡. Christine Ryan...
0 downloads 0 Views 1MB Size
pubs.acs.org/acsmedchemlett

Identification of an Orally Bioavailable, Potent, and Selective Inhibitor of GlyT1 Wesley P. Blackaby,*,†,§ Richard T. Lewis,† Joanne L. Thomson,† Andrew S. R. Jennings,† Simon C. Goodacre,† Leslie J. Street,† Angus M. MacLeod,† Andrew Pike,† Suzanne Wood,† Steve Thomas,† Terry A. Brown,† Alison Smith,† Gopalan Pillai,† Sarah Almond,† Martin R. Guscott,† H. Donald Burns,‡ Waisi Eng,‡ Christine Ryan,‡ Jacquelynn Cook,‡ and Terence G. Hamill‡ †

Merck Sharp and Dohme, Neuroscience Research Centre, Terlings Park, Eastwick Road, Harlow, Essex CM20 2QR, United Kingdom, and ‡Research Imaging, Merck Research Laboratories, West Point, Pennsylvania 19486 ABSTRACT Amalgamation of the structure-activity relationship of two series of GlyT1 inhibitors developed at Merck led to the discovery of a clinical candidate, compound 16 (DCCCyB), which demonstrated excellent in vivo occupancy of GlyT1 transporters in rhesus monkey as determined by displacement of a PET tracer ligand. KEYWORDS Inhibitor, GlyT1, structure-activity relationship, DCCCyB, PET tracer ligand

he hypofunction of N-methyl-D-aspartate (NMDA) receptors has been implicated in the pathophysiology of schizophrenia, with evidence coming from both preclinical models1-3 and the limited clinical data available.4-10 The latter includes the reversible psychosis induced in nonschizophrenics by NMDA antagonists such as phencyclidine (PCP), and the clinical efficacy observed when antipsychotic medication was supplemented with the obligatory NMDA coagonist glycine or with sarcosine, a weak endogenous inhibitor of type 1 glycine uptake transporters (GlyT1). Most importantly, this adjunctive therapy has been shown to give significant improvements in the negative and cognitive symptoms of stable schizophrenics, for which significant unmet medical need remains due to the lack of efficacy of conventional antipsychotics against these symptoms.6 In the brain, glycine levels are thought to be maintained tonically at submaximal concentrations in the synapse by GlyT1.11 This suggests that the pharmacological manipulation of synaptic glycine concentration using a GlyT1 inhibitor may be a viable method of potentiating NMDA receptor function in vivo, hence ameliorating the negative and cognitive symptoms of schizophrenia. To test this hypothesis, there has been an industry wide effort to identify potent and selective GlyT1 inhibitors.12-15 Recent communications from our laboratory have disclosed two related series of hGlyT1 inhibitors, typified by the piperidine sulfonamide 116 and cyclohexyl sulfone 2,17 which demonstrate excellent selectivity over the related GlyT2 and TauT transporters (Figure 1). Early issues for the piperidine series were poor oral bioavailability in rat and dog due to high plasma clearance, although this was resolved in the 2,4-dichlorophenyl analogue 1.

T

r 2010 American Chemical Society

However, compound 1 was shown to have unacceptably high covalent binding in vivo to both rat liver and plasma proteins after oral dosing at 20 mg/kg (6 h postdose: liver, 153 pmol equiv/mg protein; plasma, 214 pmol equiv/mg protein). Metabolite identification experiments using radiolabeled piperidine sulfonamide analogues, or trapping experiments with radiolabeled cyanide, led us to hypothesize that the observed covalent binding was due to oxidation of the piperidine ring leading to the formation of a reactive species. High levels of covalent binding have been linked to increased incidence of idiosyncratic toxicities of compounds in the clinic;18 therefore, further chemical optimization was undertaken in the sulfone series. The required cyclohexyl sulfone derivatives were accessed as delineated in Scheme 1.19 The cis and trans isomers of the key alcohol intermediate A were separated chromatographically and assigned based upon nuclear Overhauser effect NMR experiments. Where the required thiol was readily available, formation of the mesylate of A followed by displacement gave the thioether. Alternatively, the thioether was accessed via Mitsunobu chemistry with thioacetate followed by a onepot deprotection-alkylation protocol. Oxidation of the thioether with oxone gave the required final compound. The inhibition at hGlyT1 transporters and microsomal turnover in rat and human microsomes for a selection of heterocyclic sulfone compounds is given in Table 1.

Received Date: May 11, 2010 Accepted Date: June 21, 2010 Published on Web Date: June 25, 2010

350

DOI: 10.1021/ml1001085 |ACS Med. Chem. Lett. 2010, 1, 350–354

pubs.acs.org/acsmedchemlett

Table 1. hGlyT1 Potency and Human and Rat Liver Microsomal Turnover of Selected Heterocyclic Sulfone Analogues

Figure 1. Structures of hGlyT1 inhibitors. Scheme 1a

Reagents and conditions: (a) KHMDS, THF and then cyclopropylmethyl bromide. (b) LiAlH4, Et2O. (c) 2,4-Dichlorobenzoyl chloride, Hunigs base, DCM. (d) HCl(aq), THF. (e) NaBH4, EtOH. (f) Chromatographic separation of isomers. (g) MsCl, pyr and then RSNa. (h) PPh3, N2(CO2iPr)2, thioacetate. (i) LiOH, THF/H2O and then RBr. (j) Oxone, acetone/water.

a IC50 values are averages of at least two measurements. The hGlyT1a isoform was used for the assay. b Turnover of GlyT1 compounds (1 μM) in rat and human liver microsomes. All incubations were carried out at 37 °C for 15 min. Protein concentration = 0.5 mg/mL; cosolvent = 0.99% MeCN þ 0.01% DMSO. For compounds in this series, LM % turnover e40 was considered acceptable for further compound progression.

Compound 2 exhibited excellent oral bioavailability in the rat and occupied GlyT1 transporters in vivo, as adjudged by our previously reported in vivo binding assay in the rat20 using a proprietary GlyT1 radiolabel, with an Occ50 of 3.4 mg/kg.17 Analysis of the plasma and brain drug levels required to achieve Occ50 (1.2 and 0.2 μM, respectively) revealed a low brain to plasma ratio of 0.16. A similarly low brain to plasma ratio of 0.1 was determined from a 10 mg/kg oral dose of compound 3. A subsequent study in mdrla þ/þ and -/- mice determined the ratio between the brain:blood ratios of the -/- and þ/þ animals to be 8.7, suggesting compound 3 to be a P-gp substrate. Compound 2 did not inhibit common Cyp isoforms (2D6, 2C9, and 3A4: IC50 > 10 μM); however, the NH-triazole analogues 4 and 5, potential metabolites of compounds 2 and 3, respectively, proved to be extremely potent inhibitors of Cyp 2C9 (compound 5 Cyp 2C9: IC50 = 10 nM). The potent Cyp inhibition, in combination with the high plasma Occ50 due to the P-gp issue, precluded the further development of triazole analogues 2 and 3. Heterocyclic sulfone analogues in which

the pendant alkyl group was linked through carbon exhibited either increased microsomal turnover (6 and 7) or reduced potency at hGlyT1 (8 and 9). Investigation of simple alkyl sulfone derivatives related to compound 1 (Table 2) established that the structureactivity relationship (SAR) was reminiscent of the previously described 4-pyridyl piperidine series16 with a significant reduction in potency observed in the series propyl 10, ethyl 11, and methyl 12. In the alkyl sulfone series, a more stringent requirement for the cis relationship between the sulfone and the amide was observed than in the heterocyclic series, with compounds 10 and 11 demonstrating >10-fold greater potency relative to 13 and 14. Although the cyclobutylmethyl compound 15 demonstrated a loss in potency relative to propyl analogue 10, the cyclopropylmethyl compound 16 (DCCCyB) retained potency but with improved microsomal stability. Compound 16 demonstrated an acceptable level of in vivo covalent binding (